Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Front Immunol ; 13: 919489, 2022.
Article in English | MEDLINE | ID: mdl-35928820

ABSTRACT

Background: Dysregulated interleukin (IL)-6 production can be characterised by the levels present, the kinetics of its rise and its inappropriate location. Rapid, excessive IL-6 production can exacerbate tissue damage in vital organs. In this situation, therapy with an anti-IL-6 or anti-IL-6 receptor (IL-6R) monoclonal antibody, if inappropriately dosed, may be insufficient to fully block IL-6 signalling and normalise the immune response. Methods: We analysed inhibition of C-reactive protein (CRP) - a biomarker for IL-6 activity - in patients with COVID-19 or idiopathic multicentric Castleman disease (iMCD) treated with tocilizumab (anti-IL-6R) or siltuximab (anti-IL-6), respectively. We used mathematical modelling to analyse how to optimise anti-IL-6 or anti-IL-6R blockade for the high levels of IL-6 observed in these diseases. Results: IL-6 signalling was insufficiently inhibited in patients with COVID-19 or iMCD treated with standard doses of anti-IL-6 therapy. Patients whose disease worsened throughout therapy had only partial inhibition of CRP production. Our model demonstrated that, in a scenario representative of iMCD with persistent high IL-6 production not controlled by a single dose of anti-IL-6 therapy, repeated administration more effectively inhibited IL-6 activity. In a situation with rapid, high, dysregulated IL-6 production, such as severe COVID-19 or a cytokine storm, repeated daily administration of an anti-IL-6/anti-IL-6R agent, or alternating daily doses of anti-IL-6 and anti-IL-6R therapies, could neutralise IL-6 activity. Conclusion: In clinical practice, IL-6 inhibition should be individualised based on pathophysiology to achieve full blockade of CRP production. Funding: EUSA Pharma funded medical writing assistance and provided access to the phase II clinical data of siltuximab for analysis.


Subject(s)
COVID-19 Drug Treatment , Castleman Disease , C-Reactive Protein/therapeutic use , Castleman Disease/drug therapy , Cytokine Release Syndrome , Humans , Precision Medicine
4.
Stem Cell Res Ther ; 11(1): 158, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32303252

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) represent an interesting tool to improve pancreatic islet transplantation. They have immunomodulatory properties and secrete supportive proteins. However, the functional properties of MSCs vary according to many factors such as donor characteristics, tissue origin, or isolation methods. To counteract this heterogeneity, we aimed to immortalize and characterize adherent cells derived from human pancreatic islets (hISCs), using phenotypic, transcriptomic, and functional analysis. METHODS: Adherent cells derived from human islets in culture were infected with a hTERT retrovirus vector and then characterized by microarray hybridization, flow cytometry analysis, and immunofluorescence assays. Osteogenic, adipogenic, and chondrogenic differentiation as well as PBMC proliferation suppression assays were used to compare the functional abilities of hISCs and MSCs. Extracellular matrix (ECM) gene expression profile analysis was performed using the SAM (Significance Analysis of Microarrays) software, and protein expression was confirmed by western blotting. RESULTS: hISCs kept an unlimited proliferative potential. They exhibited several properties of MSCs such as CD73, CD90, and CD105 expression and differentiation capacity. From a functional point of view, hISCs inhibited the proliferation of activated peripheral blood mononuclear cells. The transcriptomic profile of hISCs highly clusterized with bone marrow (BM)-MSCs and revealed a differential enrichment of genes involved in the organization of the ECM. Indeed, the expression and secretion profiles of ECM proteins including collagens I, IV, and VI, fibronectin, and laminins, known to be expressed in abundance around and within the islets, were different between hISCs and BM-MSCs. CONCLUSION: We generated a new human cell line from pancreatic islets, with MSCs properties and retaining some pancreatic specificities related to the production of ECM proteins. hISCs appear as a very promising tool in islet transplantation by their availability (as a source of inexhaustible source of cells) and ability to secrete a supportive "pancreatic" microenvironment.


Subject(s)
Islets of Langerhans , Mesenchymal Stem Cells , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrogenesis , Humans , Leukocytes, Mononuclear
5.
Cancers (Basel) ; 11(4)2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30925767

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is the second most common hematological cancer after lymphoma. It is characterized by the accumulation of clonal malignant plasma cells within the bone marrow. The development of drug resistance remains a major problem for effective treatment of MM. Understand the mechanisms underlying drug resistance in MM is a focal point to improve MM treatment. METHODS: In the current study, we analyzed further the role of redox imbalance induction in melphalan-induced toxicity both in human myeloma cell lines (HMCLs) and primary myeloma cells from patients. RESULTS: We developed an in-vitro model of short-term resistance to high-dose melphalan and identified that pretreatment with physiological concentration of GSH protects HMCLs from melphalan-induced cell cycle arrest and cytotoxicity. We validated these results using primary MM cells from patients co-cultured with their bone marrow microenvironment. GSH did not affect the ability of melphalan to induce DNA damages in MM cells. Interestingly, melphalan induced reactive oxygen species, a significant decrease in GSH concentration, protein and lipd oxydation together with NRF2 (NF-E2-related factor 2) pathway activation. CONCLUSIONS: Our data demonstrate that antioxidant defenses confers resistance to high dose melphalan in MM cells, supporting that redox status in MM cells could be determinant for patients' response to melphalan.

6.
Theranostics ; 9(2): 540-553, 2019.
Article in English | MEDLINE | ID: mdl-30809292

ABSTRACT

Human multiple myeloma tumor cell lines (HMCLs) have been a cornerstone of research in multiple myeloma (MM) and have helped to shape our understanding of molecular processes that drive tumor progression. A comprehensive characterization of genomic mutations in HMCLs will provide a basis for choosing relevant cell line models to study a particular aspect of myeloma biology, or to screen for an antagonist of certain cancer pathways. Methods: We performed whole exome sequencing on a large cohort of 30 HMCLs, representative of a large molecular heterogeneity of MM, and 8 control samples (epstein-barr virus (EBV)-immortalized B-cells obtained from 8 different patients). We evaluated the sensitivity of HMCLs to ten drugs. Results: We identified a high confidence list of 236 protein-coding genes with mutations affecting the structure of the encoded protein. Among the most frequently mutated genes, there were known MM drivers, such as TP53, KRAS, NRAS, ATM and FAM46C, as well as novel mutated genes, including CNOT3, KMT2D, MSH3 and PMS1. We next generated a comprehensive map of altered key pathways in HMCLs. These include cell growth pathways (MAPK, JAK-STAT, PI(3)K-AKT and TP53 / cell cycle pathway), DNA repair pathway and chromatin modifiers. Importantly, our analysis highlighted a significant association between the mutation of several genes and the response to conventional drugs used in MM as well as targeted inhibitors. Conclusion: Taken together, this first comprehensive exome-wide analysis of the mutational landscape in HMCLs provides unique resources for further studies and identifies novel genes potentially associated with MM pathophysiology, some of which may be targets for future therapeutic intervention.


Subject(s)
DNA Mutational Analysis , Disease Progression , Drug Resistance, Neoplasm , Multiple Myeloma/pathology , Cell Line, Tumor , Exome , Gene Expression Regulation , Humans , Metabolic Networks and Pathways/genetics , Signal Transduction/genetics , Whole Genome Sequencing
7.
Cytometry B Clin Cytom ; 96(3): 209-214, 2019 05.
Article in English | MEDLINE | ID: mdl-30417559

ABSTRACT

BACKGROUND: Mutiple myeloma (MM) is a neoplasia characterized by the accumulation of malignant plasma cells (PC) in the bone marrow. Although proliferation markers have been studied in MM, none of the current staging systems include them. Moreover, approaches used to analyze proliferation do not separate MM cells (MMCs) from normal PC. METHODS: In this study, we combined multiparameter flow cytometry and BrdU incorporation or Ki67 staining to analyze MM cell proliferation in 44 monoclonal gammopathy of undetermined significance (MGUS), 153 newly diagnosed MM patients and 69 MM patients at relapse. The prognostic value of proliferation assessment was analyzed in 60 newly diagnosed patients treated with high-dose chemotherapy supported by autologous hematopoietic stem cell transplantation. RESULTS: The median number of proliferating malignant PC significantly increases during MM disease progression. MM patients with a percentage of proliferating MMCs greater than 1.42% using BrdU/DAPI or greater than 1.1% using ki67/DAPI, are associated with a significantly shorter event free survival compared with patients with a lower percentage of proliferating MMCs. CONCLUSIONS: Combination of flow cytometry with BrdU or ki67/DAPI staining could become a standard for the determination of MM cell proliferation. Furthermore, in the context of new effective myeloma treatment options, assessment of MM cell proliferation may be valuable, in clinical trials, to identify novel agents that could significantly affect the small proliferative compartment of MM cells. © 2018 International Clinical Cytometry Society.


Subject(s)
Flow Cytometry/methods , Hematopoietic Stem Cell Transplantation/methods , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Multiple Myeloma/diagnosis , Plasma Cells/pathology , Staining and Labeling/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bromodeoxyuridine/chemistry , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Cohort Studies , Diagnosis, Differential , Female , Humans , Indoles/chemistry , Ki-67 Antigen/metabolism , Lymphocyte Count , Male , Monoclonal Gammopathy of Undetermined Significance/mortality , Monoclonal Gammopathy of Undetermined Significance/pathology , Monoclonal Gammopathy of Undetermined Significance/therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Neoplasm, Residual , Plasma Cells/immunology , Prognosis , Progression-Free Survival , Recurrence , Transplantation, Autologous
8.
Br J Cancer ; 118(8): 1062-1073, 2018 04.
Article in English | MEDLINE | ID: mdl-29500406

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is the second most common hematologic malignancy. Aberrant epigenetic modifications have been reported in MM and could be promising therapeutic targets. As response rates are overall limited but deep responses occur, it is important to identify those patients who could indeed benefit from epigenetic-targeted therapy. METHODS: Since HDACi and DNMTi combination have potential therapeutic value in MM, we aimed to build a GEP-based score that could be useful to design future epigenetic-targeted combination trials. In addition, we investigated the changes in GEP upon HDACi/DNMTi treatment. RESULTS: We report a new gene expression-based score to predict MM cell sensitivity to the combination of DNMTi/HDACi. A high Combo score in MM patients identified a group with a worse overall survival but a higher sensitivity of their MM cells to DNMTi/HDACi therapy compared to a low Combo score. In addition, treatment with DNMTi/HDACi downregulated IRF4 and MYC expression and appeared to induce a mature BMPC plasma cell gene expression profile in myeloma cell lines. CONCLUSION: In conclusion, we developed a score for the prediction of primary MM cell sensitivity to DNMTi/HDACi and found that this combination could be beneficial in high-risk patients by targeting proliferation and inducing maturation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cellular Reprogramming/drug effects , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Multiple Myeloma/drug therapy , Plasma Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred C57BL , Microarray Analysis , Molecular Targeted Therapy/methods , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Plasma Cells/physiology , Research Design , Transcriptome , Tumor Cells, Cultured
9.
Cytometry B Clin Cytom ; 94(3): 484-492, 2018 05.
Article in English | MEDLINE | ID: mdl-28865180

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is an incurable disease characterized by clonal plasma cell (PC) proliferation within the bone marrow (BM). Next-generation flow cytometry has become the reference tool to follow minimal residual disease (MRD). We developed a new simpler and cheaper flow cytometry method to analyze bone marrow samples in patients with MM. METHODS: To identify and characterize abnormal PCs, we designed a simple panel composed of anti-CD38, antikappa, and antilambda light chain antibodies, combined with two antibody pools with the same fluorophore (anti-CD19 and anti-CD27 for the negative pool and anti-CD56, anti-CD117, and anti-CD200 antibodies for the positive pool). We also developed dedicated software for the automated identification of malignant PCs and MRD assessment. We then compared PC identification with our simple antibody panel and with the larger antibody panel routinely used at Montpellier University Hospital Center in 52 patients with MM (M-CHU cohort). RESULTS: Results for total PC detection (r2  = 0.9965; P < 0.001; n = 52) and malignant PC detection (r2  = 0.9486; P < 0.001; n = 38) obtained with the two panels were significantly correlated. Moreover, comparison of the results obtained by automated detection with our software and by manual gating analysis in 80 BM samples (38 from the M-CHU cohort and 42 patients from another MM cohort) showed strong correlation for both total and malignant PC selection (respectively, r2  = 0.936; P < 0.001 and r2  = 0.9505; P < 0.001). CONCLUSIONS: Our simple and automated strategy for MRD assessment in MM could help increasing reproducibility and productivity without compromising sensitivity and specificity, while decreasing the test cost. © 2017 International Clinical Cytometry Society.


Subject(s)
Multiple Myeloma/pathology , Plasma Cells/pathology , Antibodies/metabolism , Antigens, CD/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Cohort Studies , Flow Cytometry/methods , Humans , Multiple Myeloma/metabolism , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Plasma Cells/metabolism , Reproducibility of Results
10.
Oncotarget ; 8(58): 98931-98944, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228738

ABSTRACT

Multiple myeloma (MM) is a B cell neoplasia characterized by clonal plasma cell (PC) proliferation. Minimal residual disease monitoring by multi-parameter flow cytometry is a powerful tool for predicting treatment efficacy and MM outcome. In this study, we compared CD antigens expression between normal and malignant plasma cells to identify new potential markers to discriminate normal from malignant plasma cells, new potential therapeutic targets for monoclonal-based treatments and new prognostic factors. Nine genes were significantly overexpressed and 16 were significantly downregulated in MMC compared with BMPC (ratio ≥2; FDR CD24, CD27, CD36 and CD302) was associated with a prognostic value in two independent cohorts of patients with MM (HM cohort and TT2 cohort, n=345). The expression level of these four genes was then used to develop a CD gene risk score that classified patients in two groups with different survival (P = 2.06E-6) in the HM training cohort. The prognostic value of the CD gene risk score was validated in two independent cohorts of patients with MM (TT2 cohort and HOVON65/GMMGHD4 cohort, n=282 patients). The CD gene risk score remained a prognostic factor that separated patients in two groups with significantly different overall survival also when using publicly available data from a cohort of relapsing patients treated with bortezomib (n=188). In conclusion, the CD gene risk score allows identifying high risk patients with MM based on CD24, CD27, CD36 and CD302 expression and could represent a powerful tool for simple outcome prediction in MM.

11.
PLoS One ; 12(6): e0179793, 2017.
Article in English | MEDLINE | ID: mdl-28636654

ABSTRACT

FCRL4 is an immunoregulatory receptor that belongs to the Fc receptor-like (FCRL) family. In healthy individuals, FCRL4 is specifically expressed by memory B cells (MBCs) localized in sub-epithelial regions of lymphoid tissues. Expansion of FCRL4+ B cells has been observed in blood and other tissues in various infectious and autoimmune disorders. Currently, the mechanisms involved in pathological FCRL4+ B cell generation are actively studied, but they remain elusive. As in vivo FCRL4+ cells are difficult to access and to isolate, here we developed a culture system to generate in vitro FCRL4+ B cells from purified MBCs upon stimulation with soluble CD40 ligand and/or CpG DNA to mimic T-cell dependent and/or T-cell independent activation, respectively. After 4 days of stimulation, FCRL4+ B cells represented 17% of all generated cells. Transcriptomic and phenotypic analyses of in vitro generated FCRL4+ cells demonstrated that they were closely related to FCRL4+ tonsillar MBCs. They strongly expressed inhibitory receptor genes, as observed in exhausted FCRL4+ MBCs from blood samples of HIV-infected individuals with high viremia. In agreement, cell cycle genes were significantly downregulated and the number of cell divisions was two-fold lower in in vitro generated FCRL4+ than FCRL4- cells. Finally, due to their reduced proliferation and differentiation potential, FCRL4+ cells were less prone to differentiate into plasma cells, differently from FCRL4- cells. Our in vitro model could be of major interest for studying the biology of normal and pathological FCRL4+ cells.


Subject(s)
B-Lymphocytes/metabolism , Receptors, Fc/metabolism , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD20/metabolism , B-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/pharmacology , Down-Regulation , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Humans , Immunophenotyping , Phenotype , Receptors, Fc/genetics , Transcriptome
12.
Nucleic Acids Res ; 45(10): 5639-5652, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28459970

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that attenuate expression of their mRNA targets. Here, we developed a new method and an R package, to easily infer candidate miRNA-mRNA target interactions that could be functional during a given biological process. Using this method, we described, for the first time, a comprehensive integrated analysis of miRNAs and mRNAs during human normal plasma cell differentiation (PCD). Our results reveal 63 miRNAs with significant temporal changes in their expression during normal PCD. We derived a high-confidence network of 295 target relationships comprising 47 miRNAs and 141 targets. These relationships include new examples of miRNAs that appear to coordinately regulate multiple members of critical pathways associated with PCD. Consistent with this, we have experimentally validated a role for the miRNA-30b/c/d-mediated regulation of key PCD factors (IRF4, PRDM1, ELL2 and ARID3A). Furthermore, we found that 24 PCD stage-specific miRNAs are aberrantly overexpressed in multiple myeloma (MM) tumor plasma cells compared to their normal counterpart, suggesting that MM cells frequently acquired expression changes in miRNAs already undergoing dynamic expression modulation during normal PCD. Altogether, our analysis identifies candidate novel key miRNAs regulating networks of significance for normal PCD and malignant plasma cell biology.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Multiple Myeloma/genetics , Plasma Cells/metabolism , RNA, Messenger/genetics , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Plasma Cells/pathology , Positive Regulatory Domain I-Binding Factor 1 , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
13.
Cell Cycle ; 16(11): 1104-1117, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28463531

ABSTRACT

Plasma cells (PCs) generation occurs in hypoxic conditions in vivo, whereas the relevance of O2 pressure in PC differentiation remains unknown. Using our in vitro PC differentiation model, we investigated the role of hypoxia in PC generation. Hypoxia increases the generation of plasmablasts (PBs) starting from memory B cells, by increasing cell cycle and division number. Reactome analysis demonstrated a significant enrichment of genes involved in HIF1α and HIF2α transcription factor network, metabolism and MYC related pathways in hypoxic compared with normoxic PBs. Hypoxia-induced metabolism alteration and MYC pathway are involved in malignant PC pathophysiology. Therefore, the expression of 28 out of the 74 genes overexpressed in hypoxic PBs compared with normoxic ones was found to be associated with an adverse prognosis (event free survival and overall survival) in newly diagnosed multiple myeloma patients. According to the role of hypoxia in supporting PBs generation through cell cycle induction, c-MYC activation and metabolism alteration, it could be involved in plasma cell tumorigenesis.


Subject(s)
Plasma Cells/metabolism , Cell Cycle/genetics , Cell Hypoxia/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Multiple Myeloma/genetics
14.
Cell Cycle ; 15(14): 1919-28, 2016 07 17.
Article in English | MEDLINE | ID: mdl-27230497

ABSTRACT

A role of the transcription factor Krüppel-like factor 4 (KLF4) in the generation of mature plasma cells (PC) is unknown. Indeed, KLF4 is critical in controlling the differentiation of various cell linages, particularly monocytes and epithelial cells. KLF4 is expressed at low levels in pro-B cells and its expression increases as they mature into pre-B cells, resting naïve B cells and memory B cells. We show here that KLF4 is expressed in human bone marrow plasma cells and its function was studied using an in vitro model of differentiation of memory B cells into long lived plasma cells. KLF4 is rapidly lost when memory B cells differentiate into highly cell cycling plasmablasts, poorly cycling early plasma cells and then quiescent long-lived plasma cells. A forced expression of KLF4 in plasmablasts enhances the yield of their differentiation into early plasma cell and long lived plasma cells, by inhibiting apoptosis and upregulating previously unknown plasma cell pathways.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Plasma Cells/cytology , Plasma Cells/metabolism , Bone Marrow Cells/cytology , Caspases/metabolism , Cell Differentiation , Cell Line , Gene Expression Profiling , Humans , Kruppel-Like Factor 4
15.
Oncotarget ; 7(19): 28096-111, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27057635

ABSTRACT

Thalidomide, lenalidomide and pomalidomide have greatly improved the outcome of patients with multiple myeloma. However, their effects on plasma cells, the healthy counterpart of myeloma cells, are unknown. Here, we investigated lenalidomide effects on normal human plasma cell generation using an in vitro model. Lenalidomide inhibited the generation of pre-plasmablasts and early plasma cells, while it moderately affected plasmablast production. It also reduced the expression level of Ikaros, Aiolos, and IRF4 transcription factors, in plasmablasts and early plasma cells. This suggests that their differential sensitivity to lenalidomide is not due to a difference in Ikaros or Aiolos degradation. Lenalidomide also inhibited long-lived plasma cell generation, but did not impair their long-term survival once generated. This last observation is in agreement with the finding that lenalidomide treatment for 3-18 months did not affect the bone marrow healthy plasma cell count in allografted patients with multiple myeloma. Our findings should prompt to investigate whether lenalidomide resistance in patients with multiple myeloma could be associated with the emergence of malignant plasmablasts or long-lived plasma cells that are less sensitive to lenalidomide.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Multiple Myeloma/drug therapy , Plasma Cells/drug effects , Thalidomide/analogs & derivatives , Cells, Cultured , Hematopoietic Stem Cell Transplantation , Humans , Lenalidomide , Multiple Myeloma/pathology , Multiple Myeloma/surgery , Neoplasm, Residual/drug therapy , Thalidomide/therapeutic use
16.
Br J Cancer ; 114(5): 519-23, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26867162

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is an incurable clonal plasma cell malignancy. The constitutive expression of HIF-1α in MM suggests that inhibition of HIF-1α-mediated transcription represents an interesting target in MM. METHODS: As p300 is a crucial co-activator of hypoxia-inducible transcription, disrupting the complex HIF-1α/p300 to target HIF activity appears to be an attractive strategy. RESULTS: We reported that chetomin, an inhibitor of HIF-1α/p300 interaction, exhibits antitumour activity in human myeloma cell lines and primary MM cells from patients. CONCLUSIONS: Our data suggest that chetomin may be of clinical value in MM and especially for patients characterised by a high EP300/HIF-1α expression and a poor prognosis.


Subject(s)
Cell Proliferation/drug effects , Disulfides/pharmacology , E1A-Associated p300 Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Indole Alkaloids/pharmacology , Multiple Myeloma/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Gene Expression Profiling , Humans , Molecular Targeted Therapy , Multiple Myeloma/genetics , Prognosis
17.
Biomark Res ; 4: 3, 2016.
Article in English | MEDLINE | ID: mdl-26877874

ABSTRACT

BACKGROUND: RECQ helicase family members act as guardians of the genome to assure proper DNA metabolism in response to genotoxic stress. Hematological malignancies are characterized by genomic instability that is possibly related to underlying defects in DNA repair of genomic stability maintenance. METHODS: We have investigated the expression of RECQ helicases in different hematological malignancies and in their normal counterparts using publicly available gene expression data. Furthermore, we explored whether RECQ helicases expression could be associated with tumor progression and prognosis. RESULTS: Expression of at least one RECQ helicase family member was found significantly deregulated in all hematological malignancies investigated when compared to their normal counterparts. In addition, RECQ helicase expression was associated with a prognostic value in acute myeloid leukemia, chronic lymphocytic leukemia, lymphoma and multiple myeloma. CONCLUSION: RECQ helicase expression is deregulated in hematological malignancies compared to their normal counterparts in association with a prognostic value. Deregulation of RECQ helicases appears to play a role in tumorigenesis and represent potent therapeutic targets for synthetic lethal approaches in hematological malignancies.

18.
Oncotarget ; 7(3): 2284-96, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26497210

ABSTRACT

Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb repressive complex 2, inhibits gene expression through methylation on lysine 27 of histone H3. EZH2 regulates normal hematopoietic stem cell self-renewal and differentiation. EZH2 also controls normal B cell differentiation. EZH2 deregulation has been described in many cancer types including hematological malignancies. Specific small molecules have been recently developed to exploit the oncogenic addiction of tumor cells to EZH2. Their therapeutic potential is currently under evaluation. This review summarizes the roles of EZH2 in normal and pathologic hematological processes and recent advances in the development of EZH2 inhibitors for the personalized treatment of patients with hematological malignancies.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Hematologic Neoplasms/pathology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Polycomb Repressive Complex 2/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , B-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Epigenesis, Genetic , Ethylenediamines/pharmacology , Gene Expression Regulation , Hematologic Neoplasms/drug therapy , Histones/metabolism , Humans , Indoles/pharmacology , Methylation , Precision Medicine , Pyrazoles/pharmacology , Pyridones/pharmacology
19.
Oncotarget ; 6(36): 39165-83, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26472281

ABSTRACT

PURPOSE: microRNAs regulate gene-expression in biological and pathophysiological processes, including multiple myeloma. Here we address i) What are the number and magnitude of changes in miRNA-expression between normal plasma cells and myeloma- or MGUS-samples, and the latter two? ii) What is the biological relevance and how does miRNA-expression impact on gene-expression? iii) Is there a prognostic significance, and what is its background? EXPERIMENTAL DESIGN: Ninety-two purified myeloma-, MGUS-, normal plasma cell- and myeloma cell line-samples were investigated using miChip-arrays interrogating 559 human miRNAs. Impact on gene-expression was assessed by Affymetrix DNA-microarrays in two cohorts of myeloma patients (n = 677); chromosomal aberrations were assessed by iFISH, survival for 592 patients undergoing up-front high-dose chemotherapy. RESULTS: Compared to normal plasma cells, 67/559 miRNAs (12%) with fold changes of 4.6 to -3.1 are differentially expressed in myeloma-, 20 (3.6%) in MGUS-samples, and three (0.5%) between MGUS and myeloma. Expression of miRNAs is associated with proliferation, chromosomal aberrations, tumor mass, and gene expression-based risk-scores. This holds true for target-gene signatures of regulated mRNAs. miRNA-expression confers prognostic significance for event-free and overall survival, as do respective target-gene signatures. CONCLUSIONS: The myeloma-miRNome confers a pattern of small changes of individual miRNAs impacting on gene-expression, biological functions, and survival.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Multiple Myeloma/genetics , Cell Proliferation/genetics , Cohort Studies , Gene Expression Profiling , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Plasma Cells/physiology , Prognosis , Survival Analysis
20.
J Hematol Oncol ; 8: 10, 2015 Feb 08.
Article in English | MEDLINE | ID: mdl-25887188

ABSTRACT

BACKGROUND: Insulin like growth factor binding protein 7 (IGFBP7) is a secreted protein binding insulin like growth factor 1 (IGF-1), insulin, vascular endothelial growth factor A (VEGFA), and activin A. It antagonizes bone morphogenetic proteins and is involved in the tumour propagation of solid as well as haematological malignancies. Its role in multiple myeloma (MM) is not defined so far. We therefore aim here to investigate its prognostic and pathophysiological role in MM. METHODS: The clinical significance of IGFBP7 gene expression was investigated by gene expression profiling in two independent cohorts (n = 948) of newly-diagnosed MM patients. Methylation of the IGFBP7 promoter was analysed by pyrosequencing and treatment of MM cell lines with 5-aza-2-deoxycytidine. The impact of IGFBP7 on MM cells was studied by CCK-8 assay, BrdU assay and flow cytometry, respectively. IGFBP7 expression in bone marrow stromal cells (BMSCs) was studied by quantitative RT-PCR. For osteoblast development, immortalized and primary human BMSCs were cultured in osteogenic differentiation medium for 7-14 days in the presence of recombinant human IGFBP7 and/or activin A. RESULTS: Median IGFBP7 expression is significantly lower in CD138-purified plasma cells from individuals with MGUS and MM, compared to normal bone marrow plasma cells. IGFBP7 gene expression in MM cells is regulated by methylation, shown by pyrosequencing and exposure to demethylating agents (5-aza-2-deoxycytidine). High expression of IGFBP7 in MM cells is associated with adverse survival in two independent cohorts of 247 and 701 newly-diagnosed MM patients treated with high-dose therapy and autologous stem cell transplantation. IGFBP7 is associated with prognostically adverse chromosomal aberrations (t(4;14) and gain of 1q21), MMSET expression, and higher myeloma cell proliferation. In vitro, IGFBP7 overcomes activin A induced osteoblast suppression and promotes osteogenesis. MM cells downregulate IGFBP7 in stromal cells, possibly contributing to the osteoblast suppression found in MM. Conversely, higher IGFBP7 expression is associated with a lower probability of myeloma bone disease. CONCLUSIONS: Our data indicate that IGFBP7 expression is a marker for a specific methylation pattern in myeloma, linked to translocation t(4;14) associated MMSET expression, showing clinical features of adverse prognosis with absence of myeloma bone disease.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/biosynthesis , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Bone Diseases/etiology , Bone Diseases/pathology , Cell Line, Tumor , DNA Methylation , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Insulin-Like Growth Factor Binding Proteins/genetics , Male , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL